IL12RB1 Human, Sf9

Interleukin 12 Receptor Beta 1 Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

Receptor-Ligand Interactions

IL12RB1 binds the p40 subunit shared by IL-12 and IL-23, forming a critical interface for cytokine signaling. Structural studies reveal:

InteractionMechanismReference
IL12RB1-p40 BindingDirect engagement via charge complementarity and hydrogen bonding
IL12RB1-IL12Rβ2Forms a heterodimeric complex for high-affinity IL-12 binding
IL12RB1-IL23RPart of the IL-23 receptor complex, enabling STAT3 activation

A cryo-EM structure of IL-12 bound to IL12RB1 and IL12Rβ2 (3.75 Å resolution) confirms that IL12RB1’s N-terminal fibronectin III domain binds the p40 subunit, while IL12Rβ2 interacts with the p35 subunit .

Alternative Splicing and Isoforms

Human IL12RB1 undergoes alternative splicing, producing two isoforms:

  • Isoform 1: Full-length transmembrane protein (IL12Rβ1) critical for IL-12/IL-23 signaling.

  • Isoform 2: Truncated soluble form (lacking transmembrane domain) that enhances IL-12 responses in T cells .

Isoform 2’s function is regulated by intragenic competition between exon 9–10 splicing and exon 9b polyadenylation, modulated by hnRNP H binding near the polyA site .

Role in Immune Signaling

IL12RB1 Human, Sf9 is used to study:

  • Th1/Th17 Differentiation: IL12RB1 promotes Th1 polarization via STAT4 activation but supports Th17 differentiation in IRF1-deficient conditions .

  • Autoimmunity and Infection: Deficiencies in IL12RB1 lead to susceptibility to mycobacterial/Salmonella infections and impaired IFN-γ production .

Therapeutic Implications

Recombinant IL12RB1 variants engineered to target the p40-IL12RB1 interface show selective agonist activity, reducing NK cell activation while maintaining T cell responses. These designs aim to minimize toxicity in cancer immunotherapy .

Comparative Analysis of IL12RB1 Human, Sf9 with Other Forms

FeatureIL12RB1 Human, Sf9IL12RB1 (Native)
GlycosylationPresent (Sf9-expressed)Present (human cell-expressed)
TagC-terminal His tagNone
Purity>90% (chromatographic purification)Variable (cell lysate-dependent)
StabilityEnhanced (recombinant design)Lower (native protein)

Immunodeficiency and Disease

IL12RB1 mutations cause autosomal recessive Mendelian susceptibility to mycobacterial disease (MSMD), characterized by:

  • Key Features: Childhood-onset infections, BCG vaccination protection, incomplete penetrance .

  • Pathogenic Mutations: Nonsense, missense, and splice variants disrupt receptor function, impairing IFN-γ production .

Experimental Models

Jurkat T cells transduced with IL12RB1 Human, Sf9 demonstrate:

  • Enhanced IL-12 Responsiveness: Isoform 2 knockdown reduces IFN-γ secretion, highlighting its regulatory role .

  • Receptor Binding Affinity: IL12RB1 binds p40 with nanomolar affinity, independent of ligand context .

Product Specs

Introduction
Interleukin-12 (IL-12) is a cytokine that plays a crucial role in the immune system's response to infections and cancer. It acts as a signaling molecule that enhances the activity of specific immune cells, such as T cells and natural killer (NK) cells, which are essential for cell-mediated immunity. IL-12 stimulates these cells to produce interferon-gamma (IFN-γ), a potent antiviral and antitumor cytokine. Additionally, IL-12 promotes the differentiation of T helper cells into the Th1 subtype, which is involved in coordinating cellular immune responses.
Description
IL12RB1, a subunit of the IL-12 receptor, was expressed in Sf9 insect cells using a baculovirus expression system. This recombinant IL12RB1 protein is a single polypeptide chain with a molecular weight of 58.4 kDa. It encompasses amino acids 24-545 of the human IL12RB1 sequence and includes a 6-amino acid Histidine tag at the C-terminus. Glycosylation contributes to the observed multiple bands between 50-70 kDa on SDS-PAGE under reducing conditions. The protein was purified to a high degree using proprietary chromatographic methods.
Physical Appearance
A clear and colorless solution that has been sterilized by filtration.
Formulation

The IL12RB1 protein solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of phosphate-buffered saline (pH 7.4), 30% glycerol, 1 mM EDTA, and 0.1 mM PMSF.

Stability
For short-term storage (up to 4 weeks), the protein solution can be stored at 4°C. For extended storage, it is recommended to store the solution in aliquots at -20°C. To ensure optimal stability during long-term storage, adding a carrier protein like HSA or BSA to a final concentration of 0.1% is advisable. Repeated freezing and thawing of the protein solution should be avoided.
Purity

The purity of the IL12RB1 protein is greater than 85%, as determined by SDS-PAGE analysis.

Synonyms
Interleukin 12 Receptor, Beta 1, IL-12 Receptor Beta Component, IL-12 Receptor Subunit Beta-1, IL-12R Subunit Beta-1, IL12RB, Interleukin-12 Receptor Subunit Beta-1, Interleukin-12 Receptor Beta-1 Chain, Cluster Of Differentiation 212, CD212 Antigen, IL-12R-Beta-1, IL-12R-BETA1, IL-12RB1, CD212, IMD30, IL12R, IL12RB1.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
CRTSECCFQD PPYPDADSGS ASGPRDLRCY RISSDRYECS WQYEGPTAGV SHFLRCCLSS GRCCYFAAGS ATRLQFSDQA GVSVLYTVTL WVESWARNQT EKSPEVTLQL YNSVKYEPPL GDIKVSKLAG QLRMEWETPD NQVGAEVQFR HRTPSSPWKL GDCGPQDDDT ESCLCPLEMN VAQEFQLRRR QLGSQGSSWS KWSSPVCVPP ENPPQPQVRF SVEQLGQDGR RRLTLKEQPT QLELPEGCQG LAPGTEVTYR LQLHMLSCPC KAKATRTLHL GKMPYLSGAA YNVAVISSNQ FGPGLNQTWH IPADTHTEPV ALNISVGTNG TTMYWPARAQ SMTYCIEWQP VGQDGGLATC SLTAPQDPDP AGMATYSWSR ESGAMGQEKC YYITIFASAH PEKLTLWSTV LSTYHFGGNA SAAGTPHHVS VKNHSLDSVS VDWAPSLLST CPGVLKEYVV RCRDEDSKQV SEHPVQPTET QVTLSGLRAG VAYTVQVRAD TAWLRGVWSQ PQRFSIEVQV SDHHHHHH

Q&A

What is the molecular structure of human IL12RB1 and how does it function in immune signaling?

Human IL12RB1 encodes a type I transmembrane protein belonging to the hemopoietin receptor superfamily. It functions as a critical subunit of both the IL-12 and IL-23 receptor complexes . The protein contains an extracellular domain that binds to the IL-12p40 subunit (common to both IL-12 and IL-23), a transmembrane domain, and a cytoplasmic domain that participates in signal transduction.

In T cells, IL-12Rβ1 forms a disulfide-linked oligomer with IL-12Rβ2 to create functional IL-12 receptors, or with IL-23R to form IL-23 receptors . These complexes are essential for TH1 and TH17 cell differentiation respectively, which play crucial roles in immunity against intracellular pathogens, particularly mycobacteria . The receptor's signaling ultimately leads to the activation of STAT4 (for IL-12) or STAT3 (for IL-23) transcription factors, driving specific immune gene expression programs.

How is IL12RB1 gene expression regulated in human tissues?

IL12RB1 expression demonstrates remarkable tissue-specific regulation with a notable allele-biased pattern. Studies using primary human tissues and PBMCs have revealed that IL12RB1 expression in lung tissue and T cells is predominantly transcribed from one allele, making it an example of monoallelic or strongly allele-biased expression . This expression pattern persists even after T cell activation, suggesting it is a stable epigenetic feature rather than a transient regulatory mechanism .

The allele-biased expression has significant implications for understanding disease susceptibility, as it effectively means that in many cells, only one functional copy of IL12RB1 is actively transcribed. This regulatory pattern likely contributes to the phenotypic variability observed in patients with heterozygous IL12RB1 mutations and may influence susceptibility to mycobacterial infections even in individuals without complete IL12RB1 deficiency .

What are the key isoforms of human IL12RB1 and how do they differ functionally?

Human IL12RB1 pre-mRNA undergoes alternative processing to generate two primary isoforms with distinct structures and functions:

Isoform 1 (IL12Rβ1): The canonical form, a type I transmembrane protein that localizes to the cell surface. It contains the complete extracellular domain, transmembrane domain, and cytoplasmic tail. This isoform functions as the primary subunit of both IL-12 and IL-23 receptor complexes and positively regulates cytokine responsiveness by binding the IL-12p40 domain common to both IL-12 and IL-23 .

Isoform 2: A shorter variant that lacks the transmembrane domain due to alternative splicing incorporating exon 9b instead of exon 10. This results in a secreted protein with a distinct C-terminal sequence . Initially predicted to be non-functional or to compete with Isoform 1, experimental evidence has surprisingly shown that Isoform 2 actually potentiates IL-12 responsiveness and promotes IL-12-dependent IFNγ secretion .

The decision between producing Isoform 1 or Isoform 2 is regulated by intragenic competition between exon 9-10 splicing and exon 9b splicing/polyadenylation. This process involves an exon 9b-associated polyadenylation site and is influenced by hnRNP H binding near this site .

What experimental approaches can determine the IL12RB1 isoform expression profile in a cell population?

To characterize IL12RB1 isoform expression, researchers can employ several complementary techniques:

  • RT-PCR with isoform-specific primers: Design primer pairs that span the unique exon junctions (exon 9-10 for Isoform 1 or exon 9-9b for Isoform 2). Quantitative RT-PCR can provide relative expression levels of each isoform.

  • RNA-Seq analysis: Deep sequencing of mRNA can identify splice variants and provide quantitative data on isoform ratios. Analysis should focus on junction reads spanning the critical exon 9-10 and exon 9-9b boundaries.

  • Western blot analysis: Using antibodies targeting the N-terminal domain (common to both isoforms) versus C-terminal domain (specific to Isoform 1) can distinguish the two protein products. Isoform 1 appears at approximately 100 kDa while Isoform 2 is slightly smaller and found in different cellular fractions .

  • Subcellular fractionation: Since Isoform 1 is membrane-bound while Isoform 2 is secreted, separating membrane and soluble fractions before immunoblotting can help distinguish between these variants .

  • Flow cytometry: Surface staining detects only Isoform 1, while intracellular staining after permeabilization can detect total IL12RB1 protein, allowing inference of Isoform 2 levels.

How does IL12RB1 deficiency affect human immune responses to infections?

IL12RB1 deficiency causes an autosomal recessive disorder characterized by increased susceptibility to mycobacterial and Salmonella infections . This phenotype reveals the critical role of IL12RB1 in human antimicrobial immunity.

The immunological consequences of IL12RB1 deficiency include:

  • Impaired IL-12 signaling: Leads to defective TH1 differentiation and reduced IFNγ production by T cells and NK cells .

  • Compromised IL-23 signaling: Results in impaired TH17 responses, reducing neutrophil recruitment and IL-17-dependent antimicrobial functions .

  • Defective macrophage activation: Insufficient IFNγ production fails to properly activate macrophages, limiting their ability to kill intracellular pathogens like Mycobacterium tuberculosis .

  • Granuloma formation abnormalities: Patients often show defects in granuloma formation, a critical containment mechanism for mycobacterial infections .

  • Selective infectious susceptibility: Despite the broad roles of IL-12 and IL-23 in immunity, patients primarily show increased susceptibility to mycobacterial and Salmonella infections, with relatively normal resistance to other pathogens .

This selective vulnerability demonstrates the non-redundant role of the IL-12/IL-23 axis specifically in immunity against intracellular bacterial pathogens.

What molecular mechanisms control the alternative splicing of IL12RB1 pre-mRNA?

The alternative processing of IL12RB1 pre-mRNA into either Isoform 1 or Isoform 2 involves a sophisticated regulatory mechanism involving competition between splicing and polyadenylation events. Research has identified several key components of this regulatory system:

  • Intragenic competition: There is direct competition between the splicing of exon 9 to exon 10 (producing Isoform 1) and the splicing of exon 9 to exon 9b followed by polyadenylation (producing Isoform 2) .

  • Alternative polyadenylation site: An exon 9b-associated polyadenylation site is critical for generating Isoform 2. When this site is utilized, it leads to premature termination of transcription and inclusion of exon 9b instead of exon 10 .

  • hnRNP H binding: Heterogeneous nuclear ribonucleoprotein H (hnRNP H) binds near the regulated polyadenylation site. While this binding appears important for the processing event, studies have shown that hnRNP H is not absolutely required for exon 9b polyadenylation, suggesting additional regulatory factors are involved .

  • Tissue-specific splicing factors: Though not fully characterized, it's likely that tissue-specific expression of various splicing factors influences the Isoform 1/Isoform 2 ratio in different cell types and activation states.

Understanding these mechanisms has significant implications for potentially modulating IL12RB1 isoform expression as a therapeutic approach.

How does allele-biased expression of IL12RB1 impact immune function and disease susceptibility?

The discovery that IL12RB1 expression is predominantly monoallelic or strongly allele-biased has profound implications for understanding immune function and disease susceptibility:

  • Reduced functional reserve: Since most cells primarily express IL12RB1 from only one allele, heterozygous mutations may have greater functional impact than would be expected in a strictly biallelic expression model .

  • Variable penetrance: Allele-biased expression may explain the variable penetrance of mycobacterial susceptibility in individuals with identical IL12RB1 mutations, as the frequency of cells expressing the mutant versus wild-type allele could differ between individuals .

  • Epigenetic stability: Research shows that the extent of allele-biased expression remains stable even after T cell activation, suggesting this is a fixed epigenetic state rather than a dynamic regulatory mechanism .

  • Tissue specificity: The degree of allele bias may vary between tissues, potentially explaining why IL12RB1 deficiency manifests primarily as mycobacterial and Salmonella susceptibility rather than broader immune dysfunction .

This allele-biased expression pattern represents an important consideration for genetic diagnosis and therapeutic approaches, as conventional heterozygote/homozygote distinctions may be insufficient to predict functional consequences of IL12RB1 variants.

What is the functional significance of IL12RB1 Isoform 2 in human immune responses?

  • Enhancement of IL-12 signaling: Microarray-mediated knockdown experiments demonstrated that Isoform 2 promotes IL-12-dependent IFNγ expression in T cells .

  • Secreted modulator: Unlike Isoform 1 (a membrane-bound receptor), Isoform 2 is secreted and has a localization pattern distinct from Isoform 1, suggesting it may act as a paracrine or endocrine modulator of IL-12 responses .

  • TB resistance factor: Studies in mouse models suggest that the homolog of Isoform 2 (IL12Rβ1ΔTM) enhances resistance to extrapulmonary tuberculosis .

  • Distinct mechanism: The biochemical mechanism by which Isoform 2 potentiates IL-12 signaling remains undefined but is likely different from Isoform 1 due to its distinct localization and structure .

This functional duality of IL12RB1 gene products represents an elegant example of how alternative RNA processing can generate proteins with complementary functions from a single genetic locus, potentially providing finer regulation of cytokine responsiveness.

How do hnRNP proteins regulate IL12RB1 processing and what are the implications for immune regulation?

Heterogeneous nuclear ribonucleoproteins (hnRNPs) play critical roles in RNA processing, and hnRNP H specifically has been implicated in IL12RB1 regulation:

  • Binding proximity: hnRNP H binds near the regulated polyadenylation site in exon 9b of IL12RB1 pre-mRNA .

  • Regulatory complexity: While hnRNP H associates with this region, experimental evidence indicates it is not strictly required for exon 9b polyadenylation, suggesting a complex regulatory landscape involving multiple factors .

  • Splicing vs. polyadenylation: hnRNP H may modulate the competition between continued splicing (leading to Isoform 1) versus terminal exon definition and polyadenylation at exon 9b (leading to Isoform 2) .

  • Context-dependent regulation: The influence of hnRNP H likely depends on cellular context and the presence of other splicing factors, potentially explaining tissue- or activation-specific variations in isoform ratios.

The involvement of hnRNPs in IL12RB1 processing represents a potential target for therapeutic manipulation of IL-12 and IL-23 responses in various disease contexts, including mycobacterial infections, autoimmunity, and cancer.

What are the most significant IL12RB1 mutations associated with mycobacterial susceptibility?

IL12RB1 deficiency is characterized by a diverse spectrum of mutations that abolish receptor function and increase susceptibility to mycobacterial infections:

Understanding these mutations provides important insights for genetic diagnosis and counseling, as well as for developing potential therapeutic approaches targeting specific mutation types.

What are the optimal strategies for expressing functional human IL12RB1 in Sf9 insect cells?

Expressing functional human IL12RB1 in Sf9 cells requires careful optimization to preserve structural integrity and post-translational modifications. Based on general principles for membrane protein expression in insect cells, the following strategies are recommended:

  • Vector selection: Baculovirus expression vectors containing strong promoters (polyhedrin or p10) are typically used. Consider vectors with secretion signals (such as gp67 or honeybee melittin) for the extracellular domain if expressing just this portion.

  • Construct design options:

    • Full-length IL12RB1 (Isoform 1) with native signal peptide

    • Soluble extracellular domain only (similar to natural Isoform 2)

    • Addition of purification tags (His, FLAG, etc.) at N- or C-terminus

    • Inclusion of TEV or other protease cleavage sites for tag removal

  • Expression optimization:

    • Test multiple MOIs (multiplicity of infection) to determine optimal virus:cell ratio

    • Optimize harvest time (typically 48-72 hours post-infection) by monitoring expression time course

    • Consider lower temperature incubation (27°C instead of 28-30°C) to slow protein production and improve folding

    • Supplement media with protease inhibitors to minimize degradation

  • Protein folding considerations:

    • Monitor glycosylation pattern as indicator of proper folding

    • Consider co-expression with human chaperones to facilitate correct folding

    • Test different cell lysis and solubilization methods to preserve native conformation

Success of expression should be verified through Western blotting, functional binding assays with IL-12, and structural analyses to confirm proper folding.

How can researchers optimize solubilization and purification of membrane-bound IL12RB1 from Sf9 cells?

Purifying transmembrane proteins like IL12RB1 (Isoform 1) from Sf9 cells requires careful membrane solubilization and chromatography strategies:

  • Cell lysis and membrane preparation:

    • Harvest cells 48-72 hours post-infection

    • Lyse cells using gentle methods (nitrogen cavitation or Dounce homogenization)

    • Separate membrane fraction through ultracentrifugation (typically 100,000×g)

    • Wash membrane pellet to remove peripheral proteins

  • Detergent screening and solubilization:

    • Test multiple detergents systematically (DDM, LMNG, Digitonin, GDN)

    • Optimize detergent:protein ratio and solubilization time/temperature

    • Consider detergent mixtures for improved stability

    • Include stabilizers (glycerol, specific lipids, cholesterol) if needed

  • Affinity chromatography:

    • Use immobilized metal affinity chromatography (IMAC) for His-tagged constructs

    • Consider antibody-based affinity purification for native protein

    • Maintain detergent concentration above CMC in all buffers

    • Elute with imidazole gradient or specific competitors

  • Secondary purification:

    • Size exclusion chromatography to separate monomers from aggregates

    • Ion exchange chromatography for removing contaminants

    • Ligand affinity chromatography (using immobilized IL-12) for functional verification

  • Quality assessment:

    • SDS-PAGE and Western blotting

    • Dynamic light scattering to assess homogeneity

    • Circular dichroism to verify secondary structure

    • Thermal stability assays to optimize buffer conditions

Each step requires optimization for the specific construct and intended application, with particular attention to maintaining the native conformation of IL12RB1.

What are the key differences between producing soluble IL12RB1 (Isoform 2) versus membrane-bound IL12RB1 (Isoform 1) in Sf9 cells?

Producing the two different isoforms of IL12RB1 in Sf9 cells presents distinct challenges and opportunities:

ParameterIsoform 1 (Membrane-bound)Isoform 2 (Soluble)
Signal peptideRequires native or heterologous signal sequenceSame requirement as Isoform 1
Construct designComplete coding sequence including transmembrane domainSequence corresponding to exons 1-9b or engineered truncation before TM domain
LocalizationCell membrane-associatedSecreted into culture medium
Harvest methodCell lysis and membrane preparationCollection of culture supernatant
Primary challengesDetergent solubilization, maintaining native conformationPotential proteolytic degradation in medium
Purification approachDetergent-based membrane protein purificationConventional secreted protein purification
Typical yieldsLower (1-5 mg/L culture)Higher (5-20 mg/L culture)
Functional validationRequires reconstitution in membranes or nanodiscsDirect binding assays in solution

The choice between producing Isoform 1 or Isoform 2 depends on the specific research questions. Isoform 2 is generally easier to produce in high quantities and may be sufficient for binding studies, while Isoform 1 is essential for structural studies of the complete receptor and for functional reconstitution experiments.

How can researchers verify that recombinant IL12RB1 produced in Sf9 cells is properly folded and functional?

Verifying the structural integrity and functionality of recombinant IL12RB1 requires multiple complementary approaches:

  • Biochemical characterization:

    • SDS-PAGE under reducing and non-reducing conditions to assess disulfide bond formation

    • Western blotting with conformation-specific antibodies

    • Mass spectrometry to confirm glycosylation pattern (IL12RB1 has multiple N-glycosylation sites)

    • Limited proteolysis to probe tertiary structure

  • Biophysical analyses:

    • Circular dichroism spectroscopy to assess secondary structure elements

    • Thermal shift assays to evaluate protein stability

    • Size exclusion chromatography coupled with multi-angle light scattering (SEC-MALS) to determine oligomeric state

    • Differential scanning calorimetry to assess domain folding

  • Functional binding assays:

    • Surface plasmon resonance (SPR) to measure binding kinetics with IL-12 and IL-23

    • Bio-layer interferometry as an alternative to SPR

    • ELISA-based binding assays

    • Co-immunoprecipitation with binding partners

  • Reconstitution experiments:

    • For Isoform 1, reconstitution into liposomes or nanodiscs

    • Co-expression with IL12RB2 to form complete receptor complexes

    • Cell-based assays using Sf9 cells expressing both receptor components

  • Functional activity tests:

    • STAT4 phosphorylation assays when co-expressed with signaling components

    • For Isoform 2, testing ability to enhance IL-12-dependent IFNγ production in appropriate bioassays

These methods collectively provide a comprehensive assessment of whether the recombinant IL12RB1 retains native-like properties and functional capabilities.

What are the challenges in co-expressing IL12RB1 with its partner chains (IL12RB2 or IL23R) in Sf9 cells?

Co-expressing IL12RB1 with its partner receptor chains presents several challenges that must be addressed for successful complex formation:

  • Vector design considerations:

    • Dual promoter vectors versus co-infection with multiple viruses

    • Balancing expression levels of partner chains for optimal complex formation

    • Incorporating different purification tags on each chain for complex verification

  • Expression timing optimization:

    • Synchronizing expression of multiple proteins

    • Determining optimal harvest time that accommodates both proteins' expression kinetics

  • Complex stability issues:

    • Identifying conditions that maintain receptor-receptor interactions

    • Selecting detergents that preserve complex integrity for membrane proteins

    • Determining whether stabilizing mutations or fusion constructs are needed

  • Purification strategy complexity:

    • Sequential affinity purification steps targeting different components

    • Native gel electrophoresis to verify complex formation

    • Size exclusion chromatography to separate fully assembled complexes from individual components

  • Functional validation challenges:

    • Confirming correct stoichiometry of assembled complexes

    • Verifying enhanced ligand binding compared to individual components

    • Testing signaling capability in reconstituted systems

A successful co-expression strategy might involve creating a dual promoter baculovirus vector expressing IL12RB1 with a His tag and IL12RB2 or IL23R with a different tag (e.g., FLAG or Strep-tag), followed by tandem affinity purification to isolate only the correctly assembled heterodimeric complexes.

What cell-based assays can assess IL12RB1 function in research settings?

Several cell-based assay systems can effectively evaluate IL12RB1 functionality:

  • STAT phosphorylation assays:

    • Measure STAT4 phosphorylation in response to IL-12 stimulation

    • Analyze STAT3 phosphorylation for IL-23 signaling

    • Can be performed using flow cytometry, Western blotting, or ELISA-based methods

    • Particularly useful for comparing wild-type versus mutant IL12RB1 variants

  • Reporter cell assays:

    • Engineer cells with STAT-responsive promoters driving luciferase/GFP expression

    • Transfect with IL12RB1 constructs (wild-type or variants)

    • Quantify reporter activation following cytokine stimulation

    • Provides sensitive readout of signaling functionality

  • Cytokine production assays:

    • Measure IFNγ secretion in response to IL-12 stimulation

    • Quantify IL-17 production for IL-23 pathway assessment

    • Can use ELISA, intracellular cytokine staining, or ELISPOT methods

    • Microarray-mediated knockdown of specific isoforms can determine their contribution

  • Proliferation assays:

    • Assess IL-12/IL-23-dependent proliferation in relevant cell types

    • Compare responses with/without IL12RB1 expression

    • Useful for functional complementation studies in receptor-deficient cells

  • Ligand binding assays:

    • Measure binding of fluorescently-labeled IL-12/IL-23 to cells expressing IL12RB1

    • Flow cytometry-based approach to quantify surface receptor expression and function

    • Can determine binding affinities and kinetics in live cells

These assays, particularly when performed in IL12RB1-deficient cells reconstituted with various constructs, provide comprehensive assessment of receptor functionality.

How can CRISPR-Cas9 be used to study IL12RB1 isoform-specific functions?

CRISPR-Cas9 genome editing offers powerful approaches to dissect isoform-specific functions of IL12RB1:

  • Isoform-specific knockout strategies:

    • Target exon 9b to specifically disrupt Isoform 2 production

    • Design guide RNAs targeting the exon 9-10 junction to selectively affect Isoform 1

    • Create complete IL12RB1 knockout as control by targeting early constitutive exons

  • Splice site modification:

    • Edit specific splice donor/acceptor sites to alter splicing patterns

    • Modify the exon 9b polyadenylation signal to affect Isoform 2 production

    • Introduce subtle mutations that affect hnRNP H binding sites

  • Allele-specific modifications:

    • Create heterozygous cell models with one wild-type and one modified allele

    • Study the effects of allele-biased expression by introducing SNPs that affect allelic expression

    • Combine with allele-specific RNA-seq to correlate genetic modifications with expression patterns

  • Reporter knock-in approaches:

    • Insert fluorescent protein tags to visualize isoform expression patterns

    • Create split reporter systems that activate only when specific splicing events occur

    • Develop dual-color systems to simultaneously track both isoforms

  • Base editing applications:

    • Use cytosine or adenine base editors to create precise point mutations

    • Introduce subtle changes to regulatory elements controlling isoform choice

    • Recreate patient-specific mutations to study their functional consequences

These approaches, particularly when applied in primary human T cells or relevant cell lines, can provide unprecedented insights into the specific contributions of each IL12RB1 isoform to immune function.

What RNA-based methods can effectively modulate IL12RB1 isoform ratios for functional studies?

Several RNA-based techniques can specifically alter IL12RB1 isoform expression patterns:

  • Isoform-specific microRNAs:

    • Design microRNAs targeting exon 9b (specific to Isoform 2) or the exon 9-10 junction (specific to Isoform 1)

    • Validate knockdown specificity through RT-PCR and Western blotting

    • Assess functional consequences through IL-12 response assays

    • This approach has been successfully used to demonstrate Isoform 2's role in enhancing IL-12-dependent IFNγ expression

  • Morpholino antisense oligonucleotides:

    • Target specific splice junctions to modify splicing patterns

    • Design morpholinos that mask the exon 9b polyadenylation signal

    • Use vivo-morpholinos for enhanced cellular uptake

  • Splice-switching oligonucleotides (SSOs):

    • Design SSOs targeting specific exonic splicing enhancers or silencers

    • Modify the competition between exon 9-10 splicing and exon 9b inclusion

    • Optimize chemistry (2'-O-methyl, LNA, phosphorothioate) for stability and efficacy

  • CRISPR-Cas13 RNA targeting:

    • Use RNA-targeting CRISPR systems to degrade specific isoform transcripts

    • Design guide RNAs recognizing unique regions of each isoform

    • Combine with dCas13 effectors to modulate splicing without degradation

  • Overexpression of splicing regulators:

    • Modulate levels of hnRNP H and other splicing factors that influence IL12RB1 processing

    • Create dominant-negative versions of these proteins to interfere with normal processing

    • Use inducible expression systems for temporal control of splicing regulation

These methods provide complementary approaches to interrogate the specific functions of IL12RB1 isoforms and the mechanisms controlling their expression ratios.

How can researchers investigate the biochemical mechanism through which IL12RB1 Isoform 2 enhances IL-12 signaling?

Investigating the mechanism of Isoform 2's enhancement of IL-12 signaling requires multiple experimental approaches:

  • Structure-function analysis:

    • Generate truncation and point mutants of Isoform 2

    • Identify domains required for enhancing activity

    • Determine if the unique C-terminal sequence resulting from exon 9b is functionally important

    • Create domain-swapping chimeras between Isoforms 1 and 2

  • Interaction studies:

    • Perform pull-down assays to identify binding partners specific to Isoform 2

    • Use biochemical cross-linking followed by mass spectrometry

    • Conduct yeast two-hybrid or BioID proximity labeling experiments

    • Determine if Isoform 2 directly interacts with IL-12, IL12RB2, or downstream signaling components

  • Localization and trafficking analysis:

    • Track secretion and localization of fluorescently-tagged Isoform 2

    • Determine if Isoform 2 modifies surface expression or turnover of IL12RB2

    • Investigate whether Isoform 2 affects receptor complex formation or stability

  • Signaling pathway assessment:

    • Compare phosphorylation kinetics of STAT4 and other signaling molecules

    • Conduct phosphoproteomics to identify unique signaling events induced by Isoform 2

    • Investigate whether Isoform 2 affects receptor internalization or recycling

    • Determine if Isoform 2 prolongs signaling duration or alters signal amplitude

  • Competitive binding studies:

    • Measure whether Isoform 2 alters IL-12 binding affinity to cell surface receptors

    • Test if Isoform 2 functions as a co-receptor or stabilizes ligand-receptor interactions

    • Determine stoichiometry of any complexes formed

These approaches should be conducted in parallel with functional readouts such as IFNγ production to correlate biochemical findings with functional outcomes .

What are the best approaches for studying allele-specific expression of IL12RB1 in primary human cells?

Investigating allele-biased expression of IL12RB1 in primary human cells requires specialized techniques:

  • SNP-based allele discrimination:

    • Identify heterozygous coding SNPs in donor samples

    • Perform allele-specific qRT-PCR using probes targeting these SNPs

    • Compare genomic DNA and cDNA to quantify allelic imbalance

    • This approach has been used to demonstrate that lung and T cell IL12RB1 expression is allele-biased

  • RNA-seq with allelic resolution:

    • Conduct deep RNA sequencing of cells from heterozygous individuals

    • Analyze read counts aligning to heterozygous SNPs

    • Use computational pipelines specifically designed for allele-specific expression analysis

    • Connect with epigenetic data to identify regulatory mechanisms

  • Single-cell approaches:

    • Perform single-cell RNA-seq to determine if allele choice varies between individual cells

    • Use single-molecule RNA FISH with allele-specific probes to visualize expression in intact cells

    • Combine with phenotypic readouts to correlate allele choice with functional outcomes

  • Epigenetic profiling:

    • Conduct ChIP-seq for histone modifications associated with active/inactive alleles

    • Perform ATAC-seq to assess chromatin accessibility at each allele

    • Analyze DNA methylation patterns using bisulfite sequencing with allelic resolution

    • These approaches can help identify mechanisms maintaining allele-biased expression

  • Longitudinal analysis:

    • Track allelic expression patterns before and after cell activation

    • Determine stability of allele choice during differentiation processes

    • Assess whether environmental signals can modify allelic imbalance

These techniques collectively provide comprehensive insights into the establishment, maintenance, and functional consequences of allele-biased IL12RB1 expression in human immunity.

Product Science Overview

Introduction

Interleukin 12 Receptor Beta 1 (IL-12Rβ1) is a crucial component of the immune system, playing a significant role in the signaling pathways of interleukin-12 (IL-12) and interleukin-23 (IL-23). This receptor is essential for the activation and differentiation of T cells and natural killer (NK) cells, which are vital for the body’s defense against infections and cancer.

Structure and Function

IL-12Rβ1 is a glycoprotein that forms part of the IL-12 receptor complex. It binds to IL-12 with low affinity and, in association with IL-12Rβ2, forms a high-affinity receptor for IL-12 . This receptor complex is involved in the signal transduction pathways that lead to the activation of Janus family tyrosine kinase 2 (JAK2) and signal transducer and activator of transcription 4 (STAT4), which are critical for the immune response .

The structure of IL-12Rβ1 has been elucidated through various techniques, including X-ray crystallography and cryoelectron microscopy. These studies have revealed the detailed interactions between IL-12Rβ1 and its ligands, providing insights into its function and potential therapeutic targeting .

Recombinant Production in Sf9 Cells

The recombinant production of IL-12Rβ1 in Sf9 cells involves the use of the baculovirus expression vector system (BEVS). Sf9 cells, derived from the ovarian tissue of the fall armyworm (Spodoptera frugiperda), are commonly used for this purpose due to their ability to produce high yields of recombinant proteins . These cells can be cultured in suspension or as adherent cultures and can grow in serum-free media, making them suitable for large-scale protein production .

The process begins with the insertion of the IL-12Rβ1 gene into the baculovirus genome, which is then used to infect Sf9 cells. The infected cells produce the recombinant IL-12Rβ1 protein, which can be harvested and purified for research and therapeutic applications .

Applications and Significance

Recombinant IL-12Rβ1 produced in Sf9 cells is used in various research and clinical applications. It is instrumental in studying the signaling pathways of IL-12 and IL-23, which are involved in immune responses against pathogens and tumors . Additionally, understanding the structure and function of IL-12Rβ1 can aid in the development of targeted therapies for autoimmune diseases and cancers .

The ability to produce recombinant IL-12Rβ1 in large quantities also facilitates the development of diagnostic tools and therapeutic agents. For instance, IL-12Rβ1 can be used to screen for potential inhibitors that may modulate the immune response, providing new avenues for treating inflammatory and autoimmune conditions .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.